837 PSA > 20NG/ML DOES NOT RULE OUT RADICAL PROSTATECTOMY IN HIGH RISK PATIENTS FOLLOWING NCCN CRITERIA ATTENDING METASTATIC PROGRESSION FREE SURVIVAL

2010 ◽  
Vol 9 (2) ◽  
pp. 267
Author(s):  
J. Rubio-Briones ◽  
I. Iborra ◽  
A. Collado ◽  
A. Gómez-Ferrer ◽  
J. Casanova ◽  
...  
2021 ◽  
Vol 22 (3) ◽  
pp. 1075
Author(s):  
Luca Bedon ◽  
Michele Dal Bo ◽  
Monica Mossenta ◽  
Davide Busato ◽  
Giuseppe Toffoli ◽  
...  

Although extensive advancements have been made in treatment against hepatocellular carcinoma (HCC), the prognosis of HCC patients remains unsatisfied. It is now clearly established that extensive epigenetic changes act as a driver in human tumors. This study exploits HCC epigenetic deregulation to define a novel prognostic model for monitoring the progression of HCC. We analyzed the genome-wide DNA methylation profile of 374 primary tumor specimens using the Illumina 450 K array data from The Cancer Genome Atlas. We initially used a novel combination of Machine Learning algorithms (Recursive Features Selection, Boruta) to capture early tumor progression features. The subsets of probes obtained were used to train and validate Random Forest models to predict a Progression Free Survival greater or less than 6 months. The model based on 34 epigenetic probes showed the best performance, scoring 0.80 accuracy and 0.51 Matthews Correlation Coefficient on testset. Then, we generated and validated a progression signature based on 4 methylation probes capable of stratifying HCC patients at high and low risk of progression. Survival analysis showed that high risk patients are characterized by a poorer progression free survival compared to low risk patients. Moreover, decision curve analysis confirmed the strength of this predictive tool over conventional clinical parameters. Functional enrichment analysis highlighted that high risk patients differentiated themselves by the upregulation of proliferative pathways. Ultimately, we propose the oncogenic MCM2 gene as a methylation-driven gene of which the representative epigenetic markers could serve both as predictive and prognostic markers. Briefly, our work provides several potential HCC progression epigenetic biomarkers as well as a new signature that may enhance patients surveillance and advances in personalized treatment.


2013 ◽  
Vol 189 (4S) ◽  
Author(s):  
Andreas Becker ◽  
Pierre Tennstedt ◽  
Thomas Steuber ◽  
Georg Salomon ◽  
Alexander Haese ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (17) ◽  
pp. 4547-4553 ◽  
Author(s):  
María-Victoria Mateos ◽  
Norma C. Gutiérrez ◽  
María-Luisa Martín-Ramos ◽  
Bruno Paiva ◽  
María-Angeles Montalbán ◽  
...  

Abstract Cytogenetic abnormalities (CAs) such as t(4;14), t(14;16) or del(17p), and nonhyperdiploidy are associated with poor prognosis in multiple myeloma. We evaluated the influence of CAs by FISH and DNA ploidy by flow cytometry on response and survival in 232 elderly, newly diagnosed multiple myeloma patients receiving an induction with weekly bortezomib followed by maintenance therapy with bortezomib-based combinations. Response was similar in the high-risk and standard-risk CA groups, both after induction (21% vs 27% complete responses [CRs]) and maintenance (39% vs 45% CR). However, high-risk patients showed shorter progression-free survival (PFS) than standard-risk patients, both from the first (24 vs 33 months; P = .04) and second randomization (17 vs 27 months; P = .01). This also translated into shorter overall survival (OS) for high-risk patients (3-year OS: 55% vs 77%; P = .001). This adverse prognosis applied to either t(4;14) or del(17p). Concerning DNA ploidy, hyperdiploid patients showed longer OS than nonhyperdiploid patients (77% vs 63% at 3 years; P = .04), and this was more evident in patients treated with bortezomib, thalidomide, and prednisone (77% vs 53% at 3 years; P = .02). The present schema does not overcome the negative prognosis of high-risk CAs and nonhyperdiploidy. This trial was registered with www.ClinicalTrials.gov as NCT00443235.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1150-1150 ◽  
Author(s):  
Maria-Victoria Mateos ◽  
Jane Estell ◽  
Wolney Barreto ◽  
Paolo Corradini ◽  
Chang-Ki Min ◽  
...  

Abstract Introduction: Daratumumab is a first-in-class monoclonal antibody targeting the plasma cell antigen, CD38, that demonstrated significant activity in combination with bortezomib and dexamethasone (DVd) compared with bortezomib and dexamethasone alone (Vd) in a pre-specified interim analysis of CASTOR, a randomized phase 3 study of relapsed or refractory multiple myeloma (RRMM; Palumbo A. N Engl J Med 2016; in press). In this abstract, we report on subgroup analyses according to the number of prior lines of therapy received, and include outcomes based on cytogenetic status, in order to identify the patient population who derived the most benefit from the DVd combination. Methods: Eligible patients (pts) received at least 1 prior line of therapy and were randomized (1:1) to 8 cycles (q3w) of Vd with or without daratumumab (16 mg/kg IV qw in Cycles 1-3, Day 1 of Cycles 4-8, then q4w until progression). For each cycle, bortezomib 1.3 mg/m2 SC was administered on Days 1, 4, 8, and 11, and dexamethasone 20 mg PO was given on Days 1, 2, 4, 5, 8, 9, 11, 12. Progression-free survival (PFS) was the primary endpoint. Numbers of prior lines of therapy were determined by the site investigator according to the IMWG consensus guidelines. High-risk cytogenetic status based on local laboratory assessments was defined as having at least one of the following abnormalities via karyotyping or fluorescence in-situ hybridization: del17p, t(4;14), or t(14;16). Minimal residual disease (MRD) was evaluated on bone marrow aspirate samples that had been prepared with Ficoll using three different thresholds (10-4, 10-5, and 10-6) based on the ClonoSEQ assay (Adaptive Biotechnologies, Seattle, WA, USA). Results: Median follow-up was 7.4 months. In the 1 to 3 prior lines (1-3 PL) subgroup (DVd, n=229; Vd, n=219), PFS was significantly longer with DVd vs Vd (median: not reached [NR] vs 7.3 mo; HR, 0.39; 95% CI, 0.28-0.55; P<0.0001); estimated 12-month PFS rates were 62.2% vs 29.3%, respectively. Median time to progression (TTP) among 1-3 PL pts was NR vs 7.4 months, respectively (HR, 0.29; 95% CI, 0.20-0.43; P<0.0001). Overall response rate (ORR) was significantly higher with DVd vs Vd (84% vs 67%; P<0.0001) and was associated with higher rates of very good partial response (VGPR) or better (62% vs 32%; P<0.0001). Among 1-3 PL pts with standard-risk cytogenetic status, PFS was significantly prolonged in DVd vs Vd (HR, 0.38; 95% CI, 0.25-0.58; P<0.0001), and estimated 12-month PFS rates were 58.7% vs 27.0%, respectively. PFS was also significantly longer in pts with high-risk cytogenetics who received DVd vs Vd (HR, 0.46; 95% CI, 0.22-0.97; P=0.0367; Figure), with estimated 12-month PFS rates of 63.2% vs 26.7%, respectively. Lastly, the rate of MRD-negative patients was significantly higher (4 fold or greater) at all evaluated thresholds (10-4, 10-5, and 10-6) among the 1-3 PL subgroup. In the 1 prior line (1 PL) subgroup (DVd, n=122; Vd, n=113), PFS was also significantly improved for DVd vs Vd (median: NR vs 7.5 mo; HR, 0.31; 95% CI, 0.18-0.52; P<0.0001); estimated 12-month PFS rates were 77.5% vs 29.4%, respectively. Median TTP among 1 PL pts was NR vs 8.4 months, respectively (HR, 0.28; 95% CI, 0.16-0.49; P<0.0001). ORR in pts with 1 PL was significantly higher for DVd vs Vd (89% vs 74%; P=0.0029), including higher rates of VGPR or better being observed for DVd vs Vd (71% vs 42%; P<0.0001). Updated efficacy and safety data, including analyses of MRD status across different sensitivity thresholds (10-4, 10-5, and 10-6), will be presented at the meeting. Conclusions: The treatment benefit of DVd vs Vd was maintained across 1-3 PL and 1 PL pts. Responses for DVd were deep and durable and correlated with significantly improved outcomes vs Vd. Despite small group sizes, treatment benefit of DVd vs Vd was also observed in high-risk cytogenetic pts receiving 1-3 PL. These data continue to support the addition of daratumumab to a standard-of-care regimen in RRMM. Figure Progression-free Survival in High-risk Patients (Defined by Cytogenetic Abnormalities) who Received 1 to 3 Prior Lines of Therapy Figure. Progression-free Survival in High-risk Patients (Defined by Cytogenetic Abnormalities) who Received 1 to 3 Prior Lines of Therapy Disclosures Mateos: Janssen, Celgene, Amgen, Takeda, BMS: Honoraria. Estell:Janssen: Membership on an entity's Board of Directors or advisory committees. Corradini:Celgene, Janssen, Novartis, Roche, Takeda, Sanofi, Servier, Gilead: Honoraria; Takeda: Consultancy. Medvedova:Oregon Health and Science University: Employment. Qi:Janssen: Employment. Schecter:Janssen: Employment, Equity Ownership. Amin:Janssen: Employment. Qin:Janssen: Employment. Deraedt:Janssen: Employment; Johnson & Johnson: Equity Ownership. Casneuf:Johnson & Johnson: Equity Ownership; Janssen R&D, Beerse, Belgium: Employment. Chiu:Janssen: Employment. Sasser:Janssen: Employment; Johnson & Johnson: Equity Ownership. Nooka:Amgen, Novartis, Spectrum: Consultancy.


2005 ◽  
Vol 173 (4S) ◽  
pp. 436-436
Author(s):  
Christopher J. Kane ◽  
Martha K. Terris ◽  
William J. Aronson ◽  
Joseph C. Presti ◽  
Christopher L. Amling ◽  
...  

Cancers ◽  
2021 ◽  
Vol 13 (13) ◽  
pp. 3274
Author(s):  
Vinson Wai-Shun Chan ◽  
Wei Shen Tan ◽  
Aqua Asif ◽  
Alexander Ng ◽  
Olayinka Gbolahan ◽  
...  

External factors, such as the coronavirus disease 2019 (COVID-19), can lead to cancellations and backlogs of cancer surgeries. The effects of these delays are unclear. This study summarised the evidence surrounding expectant management, delay radical prostatectomy (RP), and neoadjuvant hormone therapy (NHT) compared to immediate RP. MEDLINE and EMBASE was searched for randomised controlled trials (RCTs) and non-randomised controlled studies pertaining to the review question. Risks of biases (RoB) were evaluated using the RoB 2.0 tool and the Newcastle–Ottawa Scale. A total of 57 studies were included. Meta-analysis of four RCTs found overall survival and cancer-specific survival were significantly worsened amongst intermediate-risk patients undergoing active monitoring, observation, or watchful waiting but not in low- and high-risk patients. Evidence from 33 observational studies comparing delayed RP and immediate RP is contradictory. However, conservative estimates of delays over 5 months, 4 months, and 30 days for low-risk, intermediate-risk, and high-risk patients, respectively, have been associated with significantly worse pathological and oncological outcomes in individual studies. In 11 RCTs, a 3-month course of NHT has been shown to improve pathological outcomes in most patients, but its effect on oncological outcomes is apparently limited.


2019 ◽  
Vol 19 (5) ◽  
pp. 363-369
Author(s):  
Ashley Albert ◽  
Sophy Mangana ◽  
Mary R. Nittala ◽  
Toms Vengaloor Thomas ◽  
Lacey Weatherall ◽  
...  

2011 ◽  
Vol 96 (10) ◽  
pp. 3217-3225 ◽  
Author(s):  
Joanna Klubo-Gwiezdzinska ◽  
Douglas Van Nostrand ◽  
Frank Atkins ◽  
Kenneth Burman ◽  
Jacqueline Jonklaas ◽  
...  

Abstract Background: The optimal management of high-risk patients with differentiated thyroid cancer (DTC) consists of thyroidectomy followed by radioiodine (131I) therapy. The prescribed activity of 131I can be determined using two approaches: 1) empiric prescribed activity of 131I (E-Rx); and 2) dosimetry-based prescribed activity of 131I (D-Rx). Aim: The aim of the study was to compare the relative treatment efficacy and side effects of D-Rx vs. E-Rx. Methods: A retrospective analysis was performed of patients with distant metastases and/or locoregionally advanced radioiodine-avid DTC who were treated with either D-Rx or E-Rx. Response to treatment was based on RECIST (Response Evaluation Criteria in Solid Tumors) 1.1 criteria. Results: The study group consisted of 87 patients followed for 51 ± 35 months, of whom 43 were treated with D-Rx and 44 with E-Rx. Multivariate analysis, controlling for age, gender, and status of metastases revealed that the D-Rx group tended to be 70% less likely to progress (odds ratio, 0.29; 95% confidence interval, 0.087–1.02; P = 0.052) and more likely to obtain complete response (CR) compared to the E-Rx group (odds ratio, 8.2; 95% confidence interval, 1.2–53.5; P = 0.029). There was an association in the D-Rx group between the observed CR and percentage of maximum tolerable activity given as a first treatment of 131I (P = 0.030). The advantage of D-Rx was specifically apparent in the locoregionally advanced group because CR was significantly higher in D-Rx vs. E-Rx in this group of patients (35.7 vs. 3.3%; P = 0.009). The rates of partial response, stable disease, and progression-free survival, as well as the frequency of side effects, were not significantly different between the two groups. Conclusion: Higher efficacy of D-Rx with a similar safety profile compared to E-Rx supports the rationale for employing individually prescribed activity in high-risk patients with DTC.


Sign in / Sign up

Export Citation Format

Share Document